1
|
Porteiro B, Roscam Abbing RLP, In Het Panhuis W, de Waart DR, Duijst S, Bolt I, Vogels EW, Levels JHM, Bosmans LA, Vos WG, Oude Elferink RPJ, Lutgens E, van de Graaf SFJ. Inhibition of hepatic bile salt uptake by Bulevirtide reduces atherosclerosis in Oatp1a1 -/-Ldlr -/- mice. J Lipid Res 2024; 65:100594. [PMID: 39009243 PMCID: PMC11382107 DOI: 10.1016/j.jlr.2024.100594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 06/05/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
Bile salts can strongly influence energy metabolism through systemic signaling, which can be enhanced by inhibiting the hepatic bile salt transporter Na+ taurocholate cotransporting polypeptide (NTCP), thereby delaying hepatic reuptake of bile salts to increase systemic bile salt levels. Bulevirtide is an NTCP inhibitor and was originally developed to prevent NTCP-mediated entry of Hepatitis B and D into hepatocytes. We previously demonstrated that NTCP inhibition lowers body weight, induces glucagon-like peptide-1 (GLP1) secretion, and lowers plasma cholesterol levels in murine obesity models. In humans, a genetic loss-of-function variant of NTCP has been associated with reduced plasma cholesterol levels. Here, we aimed to assess if Bulevirtide treatment attenuates atherosclerosis development by treating female Ldlr-/- mice with Bulevirtide or vehicle for 11 weeks. Since this did not result in the expected increase in plasma bile salt levels, we generated Oatp1a1-/-Ldlr-/- mice, an atherosclerosis-prone model with human-like hepatic bile salt uptake characteristics. These mice showed delayed plasma clearance of bile salts and elevated bile salt levels upon Bulevirtide treatment. At the study endpoint, Bulevirtide-treated female Oatp1a1-/-Ldlr-/- mice had reduced atherosclerotic lesion area in the aortic root that coincided with lowered plasma LDL-c levels, independent of intestinal cholesterol absorption. In conclusion, Bulevirtide, which is considered safe and is EMA-approved for the treatment of Hepatitis D, reduces atherosclerotic lesion area by reducing plasma LDL-c levels. We anticipate that its application may extend to atherosclerotic cardiovascular diseases, which warrants clinical trials.
Collapse
Affiliation(s)
- Begoña Porteiro
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands; CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Reinout L P Roscam Abbing
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Wietse In Het Panhuis
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Dirk R de Waart
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Suzanne Duijst
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Isabelle Bolt
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Esther W Vogels
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Johannes H M Levels
- Amsterdam UMC, Department of Experimental Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura A Bosmans
- Amsterdam UMC, location AMC, Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands; Amsterdam institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Winnie G Vos
- Amsterdam UMC, location AMC, Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands; Amsterdam institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands
| | - Ronald P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Lutgens
- Amsterdam UMC, location AMC, Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands; Amsterdam institute for Immunology and Infectious Diseases, Inflammatory Diseases, Amsterdam, The Netherlands; Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Center, Amsterdam, The Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Zhang L, Liu X, Jin T, Dong J, Li X, Zhang Y, Liu D. Isomers-oriented separation of forty-five plasma bile acids with liquid chromatography-tandem mass spectrometry. J Chromatogr A 2024; 1721:464827. [PMID: 38520985 DOI: 10.1016/j.chroma.2024.464827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Some bile acids (BAs) were considered as biomarkers or have therapeutical effect on metabolic diseases. However, due to the existence of isomers and limitations in sensitivity, simultaneous quantification of multiple BAs remains a challenge. The aim of this study is to establish an accurate and sensitive method for the determination of multiple BAs with similar polarity. A LC-MS/MS analytical method capable of quantifying forty-five BAs simultaneously using nine stable isotope internal standards was developed and fully validated based on key isomers-oriented separation strategy. The method was further applied to analyze plasma samples to describe the dynamic profile of BAs after high glucose intake. The chromatography and mass spectrum conditions were optimized to enable the accurate quantification of forty-five BAs, while ensuring the lower limit of quantification between 0.05-10 ng/mL. The results of system suitability, linearity, dilution integrity, accuracy and precision demonstrated the good quantitative capacity and robustness of the method. A total of thirty-five BAs were quantified in plasma samples from twelve healthy Chinese individuals. The established method featured superior sensitivity and better separation efficiency compared to previous studies. Meanwhile, BAs exhibited correlations with glucose and insulin, suggesting their potential as biomarkers for metabolic disorders.
Collapse
Affiliation(s)
- Lei Zhang
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Xu Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China
| | - Tenghui Jin
- Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Jing Dong
- Shimadzu China Innovation Center, Beijing 100020, China
| | - Xiaodong Li
- Shimadzu China Innovation Center, Beijing 100020, China
| | - Youyi Zhang
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China.
| | - Dongyang Liu
- Center of Clinical Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China; Drug Clinical Trial Center, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
3
|
Neef SK, Hofmann U, Mürdter TE, Schwab M, Haag M. Performance comparison of narrow-bore and capillary liquid-chromatography for non-targeted metabolomics profiling by HILIC-QTOF-MS. Talanta 2023; 260:124578. [PMID: 37119797 DOI: 10.1016/j.talanta.2023.124578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/13/2023] [Accepted: 04/19/2023] [Indexed: 05/01/2023]
Abstract
Clinical metabolomics studies often have to cope with limited sample amounts, thus miniaturized liquid chromatography (LC) systems are a promising alternative. Their applicability has already been demonstrated in various fields, including a few metabolomics studies that mainly used reversed-phase chromatography. However, hydrophilic interaction chromatography (HILIC), which is widely used in metabolomics due to its particular suitability for the analysis of polar molecules, has rarely been tested for miniaturized LC-MS analysis of small molecules. In the present work, the suitability of a capillary HILIC (CapHILIC)-QTOF-MS system for non-targeted metabolomics was evaluated based on extracts of porcine formalin-fixed, paraffin-embedded (FFPE) tissue samples. The performance was assessed with respect to the number and retention time span of metabolic features as well as the analytical repeatability, the signal-to-noise ratio and the signal intensity of 16 annotated metabolites from different compound classes. The results were compared with a well established narrow-bore HILIC-QTOF-MS system. Both platforms have detected a similar number of features and performed excellent with respect to retention time stability (median RT span <0.05 min) and analytical repeatability (>75% of features with CV < 20%). The signal areas of all metabolites assessed were increased up to 18-fold by the use of CapHILIC, although the signal-to-noise ratio was only improved for 50% of the metabolites. An even better reproducibility (median CV = 5.2%) and up to 80-fold increase in signal intensity were observed after optimization of CapHILIC conditions for analysis of bile acid standard solutions. Even though the observed improvement for specific bile acids (e.g. taurocholic acid) in biological matrix needs to be evaluated, the platform comparison indicates, that the tested CapHILIC system is particularly suitable for analyses of a less broad metabolite spectrum, and specifically optimized chromatography.
Collapse
Affiliation(s)
- Sylvia K Neef
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Tuebingen, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Tuebingen, Germany; Departments of Clinical Pharmacology, and of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
4
|
Zhu V, Burhenne J, Weiss J, Haag M, Hofmann U, Schwab M, Urban S, Mikus G, Czock D, Haefeli WE, Blank A. Evaluation of the drug-drug interaction potential of the novel hepatitis B and D virus entry inhibitor bulevirtide at OATP1B in healthy volunteers. Front Pharmacol 2023; 14:1128547. [PMID: 37089922 PMCID: PMC10117888 DOI: 10.3389/fphar.2023.1128547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction: Bulevirtide is a first-in-class antiviral drug to treat chronic hepatitis B/D. We investigated the drug-drug interaction potential and pharmacokinetics of high-dose subcutaneous bulevirtide (5 mg twice daily) with organic anion transporting polypeptide 1B1 (OATP1B1) and cytochrome P450 (CYP) 3A4. Methods: This was a single-center, open-label, fixed-sequence drug-drug interaction trial in 19 healthy volunteers. Before and at bulevirtide steady state, participants ingested a single 40 mg dose of pravastatin. A midazolam microdose was applied to quantify CYP3A4 activity. Results: At bulevirtide steady state, pravastatin area under the concentration-time curve (AUC0-∞) increased 1.32-fold (90% CI 1.08-1.61). The 5 mg bulevirtide twice-daily treatment resulted in a mean AUC0-12 of 1210 h*ng/ml (95% CI 1040-1408) and remained essentially unchanged under the influence of pravastatin. CYP3A4 activity did not change to a clinically relevant extent. As expected, total bile acids increased substantially (35-fold) compared to baseline during bulevirtide treatment. All study medication was well tolerated. Discussion: The study demonstrated that high-dose bulevirtide inhibited OATP1B-mediated hepatic uptake of the marker substrate pravastatin but the extent is considered clinically not relevant. Changes in CYP3A4 activity were also not clinically relevant. In conclusion, this study suggests that OATP1B substrate drugs as well as CYP3A4 substrates may safely be used without dose adjustment in patients treated with bulevirtide. However, in patients using high statin doses and where concomitant factors potentially further increase statin exposure, caution may be required when using bulevirtide.
Collapse
Affiliation(s)
- Vanessa Zhu
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Jürgen Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Johanna Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
- Departments of Clinical Pharmacology and of Biochemistry and Pharmacy, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180), Image‐guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany
| | - Stephan Urban
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - David Czock
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Walter E. Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF) Partner Site Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- *Correspondence: Antje Blank,
| |
Collapse
|
5
|
Metabolic Effect of Blocking Sodium-Taurocholate Co-Transporting Polypeptide in Hypercholesterolemic Humans with a Twelve-Week Course of Bulevirtide-An Exploratory Phase I Clinical Trial. Int J Mol Sci 2022; 23:ijms232415924. [PMID: 36555566 PMCID: PMC9787649 DOI: 10.3390/ijms232415924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/28/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Bile acids (BA) play an important role in cholesterol metabolism and possess further beneficial metabolic effects as signalling molecules. Blocking the hepatocellular uptake of BA via sodium-taurocholate co-transporting polypeptide (NTCP) with the first-in-class drug bulevirtide, we expected to observe a decrease in plasma LDL cholesterol. In this exploratory phase I clinical trial, volunteers with LDL cholesterol > 130 mg/dL but without overt atherosclerotic disease were included. Thirteen participants received bulevirtide 5 mg/d subcutaneously for 12 weeks. The primary aim was to estimate the change in LDL cholesterol after 12 weeks. Secondary endpoints included changes in total cholesterol, HDL cholesterol, lipoprotein(a), inflammatory biomarkers, and glucose after 12 weeks. In addition, cardiac magnetic resonance imaging (CMR) was performed at four time points. BA were measured as biomarkers of the inhibition of hepatocellular uptake. After 12 weeks, LDL cholesterol decreased not statistically significantly by 19.6 mg/dL [−41.8; 2.85] (Hodges−Lehmann estimator with 95% confidence interval). HDL cholesterol showed a significant increase by 5.5 mg/dL [1.00; 10.50]. Lipoprotein(a) decreased by 1.87 mg/dL [−7.65; 0]. Inflammatory biomarkers, glucose, and cardiac function were unchanged. Pre-dose total BA increased nearly five-fold (from 2026 nmol/L ± 2158 (mean ± SD) at baseline to 9922 nmol/L ± 7357 after 12 weeks of treatment). Bulevirtide was generally well tolerated, with most adverse events being administration site reactions. The exploratory nature of the trial with a limited number of participants allows the estimation of potential effects, which are crucial for future pharmacological research on bile acid metabolism in humans.
Collapse
|
6
|
Quantitative Profiling of Bile Acids in Feces of Humans and Rodents by Ultra-High-Performance Liquid Chromatography–Quadrupole Time-of-Flight Mass Spectrometry. Metabolites 2022; 12:metabo12070633. [PMID: 35888757 PMCID: PMC9323729 DOI: 10.3390/metabo12070633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
A simple, sensitive, and reliable quantification and identification method was developed and validated for simultaneous analysis of 58 bile acids (BAs) in human and rodent (mouse and rat) fecal samples. The method involves an extraction step with a 5% ammonium–ethanol aqueous solution; the BAs were quantified by high-resolution mass spectrometry (ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry, UPLC–Q-TOF). The recoveries were 80.05–120.83%, with coefficient variations (CVs) of 0.01–9.82% for three biological species. The limits of detection (LODs) were in the range of 0.01–0.24 μg/kg, and the limits of quantification (LOQs) ranged from 0.03 to 0.81 μg/kg. In addition, the analytical method was used to identify and quantify BAs in end-stage renal disease (ESRD) patients, C57BL/6 mice, and Sprague-Dawley (SD) rats. The fecal BA profile and analysis of BA indices in these samples provide valuable information for further BA metabolic disorder research.
Collapse
|
7
|
Tremmel R, Nies AT, van Eijck BAC, Handin N, Haag M, Winter S, Büttner FA, Kölz C, Klein F, Mazzola P, Hofmann U, Klein K, Hoffmann P, Nöthen MM, Gaugaz FZ, Artursson P, Schwab M, Schaeffeler E. Hepatic Expression of the Na+-Taurocholate Cotransporting Polypeptide Is Independent from Genetic Variation. Int J Mol Sci 2022; 23:ijms23137468. [PMID: 35806468 PMCID: PMC9267852 DOI: 10.3390/ijms23137468] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
The hepatic Na+-taurocholate cotransporting polypeptide NTCP/SLC10A1 is important for the uptake of bile salts and selected drugs. Its inhibition results in increased systemic bile salt concentrations. NTCP is also the entry receptor for the hepatitis B/D virus. We investigated interindividual hepatic SLC10A1/NTCP expression using various omics technologies. SLC10A1/NTCP mRNA expression/protein abundance was quantified in well-characterized 143 human livers by real-time PCR and LC-MS/MS-based targeted proteomics. Genome-wide SNP arrays and SLC10A1 next-generation sequencing were used for genomic analyses. SLC10A1 DNA methylation was assessed through MALDI-TOF MS. Transcriptomics and untargeted metabolomics (UHPLC-Q-TOF-MS) were correlated to identify NTCP-related metabolic pathways. SLC10A1 mRNA and NTCP protein levels varied 44-fold and 10.4-fold, respectively. Non-genetic factors (e.g., smoking, alcohol consumption) influenced significantly NTCP expression. Genetic variants in SLC10A1 or other genes do not explain expression variability which was validated in livers (n = 50) from The Cancer Genome Atlas. The identified two missense SLC10A1 variants did not impair transport function in transfectants. Specific CpG sites in SLC10A1 as well as single metabolic alterations and pathways (e.g., peroxisomal and bile acid synthesis) were significantly associated with expression. Inter-individual variability of NTCP expression is multifactorial with the contribution of clinical factors, DNA methylation, transcriptional regulation as well as hepatic metabolism, but not genetic variation.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Anne T. Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
| | - Barbara A. C. van Eijck
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Niklas Handin
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Florian A. Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Charlotte Kölz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Franziska Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Pascale Mazzola
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany; (P.H.); (M.M.N.)
- Division of Medical Genetics, Department of Biomedicine, University of Basel, 4001 Basel, Switzerland
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany; (P.H.); (M.M.N.)
- Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany
| | - Fabienne Z. Gaugaz
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Per Artursson
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (N.H.); (F.Z.G.); (P.A.)
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
- Departments of Clinical Pharmacology, and of Pharmacy and Biochemistry, University of Tuebingen, 72076 Tuebingen, Germany
- Correspondence: ; Tel.: +49-711-8101-3700
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany; (R.T.); (A.T.N.); (B.A.C.v.E.); (M.H.); (S.W.); (F.A.B.); (C.K.); (F.K.); (P.M.); (U.H.); (K.K.); (E.S.)
- University of Tuebingen, 72076 Tuebingen, Germany
- iFIT Cluster of Excellence (EXC2180) “Image Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
8
|
Leoni S, Casabianca A, Biagioni B, Serio I. Viral hepatitis: Innovations and expectations. World J Gastroenterol 2022; 28:517-531. [PMID: 35316960 PMCID: PMC8905017 DOI: 10.3748/wjg.v28.i5.517] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/14/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Viral hepatitis is a significant health problem worldwide, associated with morbidity and mortality. Hepatitis B, C, D, and occasionally E viruses (HBV, HCV, HDV, and HEV) can evolve in chronic infections, whereas hepatitis A virus (HAV) frequently produces acute self-limiting hepatitis. In the last years, different studies have been performed to introduce new antiviral therapies. The most important goal in the treatment of viral hepatitis is to avoid chronic liver disease and complications. This review analyzes currently available therapies, in particular for viruses associated with chronic liver disease. The focus is especially on HBV and HCV therapies, investigating new drugs already introduced in clinical practice and clinical trials. We also describe new entry inhibitors, developed for the treatment of chronic HDV and HBV and currently available treatments for HEV. The last drugs introduced have shown important efficacy in HCV, with achievable target HCV elimination by 2030. Concurrently, renewed interest in curative HBV therapies has been registered; current nucleotide/ nucleoside analogs positively impact liver-related complications, ensuring high safety and tolerability. Novel approaches to HBV cure are based on new antivirals, targeting different steps of the HBV life cycle and immune modulators. The improved knowledge of the HDV life cycle has facilitated the development of some direct-acting agents, as bulevirtide, the first drug conditionally approved in Europe for HDV associated compensated liver disease. Further studies are required to identify a new therapeutic approach in hepatitis E, especially in immunosuppressed patients.
Collapse
Affiliation(s)
- Simona Leoni
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Alberto Casabianca
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Benedetta Biagioni
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| | - Ilaria Serio
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
| |
Collapse
|
9
|
Yang F, Xu W, Wu L, Yang L, Zhu S, Wang L, Wu W, Zhang Y, Chong Y, Peng L. NTCP Deficiency Affects the Levels of Circulating Bile Acids and Induces Osteoporosis. Front Endocrinol (Lausanne) 2022; 13:898750. [PMID: 35937832 PMCID: PMC9353038 DOI: 10.3389/fendo.2022.898750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The p.Ser267Phe mutation in the SLC10A1 gene can cause NTCP deficiency. However, the full clinical presentation of p.Ser267Phe homozygous individuals and its long-term consequences remain unclear. Hence, in the present study, we characterized the phenotypic characteristics of NTCP deficiency and evaluated its long-term prognosis. METHODS Ten NTCP p.Ser267Phe homozygous individuals were recruited and a comprehensive medical evaluation with a 5-year follow-up observation was performed. The phenotypic characteristics of NTCP deficiency were also demonstrated using an NTCP-global knockout mouse model. RESULTS During the 5-year follow-up observation of 10 NTCP p.Ser267Phe homozygous adults, we found that the most common phenotypic features of NTCP deficiency in adults were hypercholanemia, vitamin D deficiency, bone loss, and gallbladder abnormalities. The profile of bile acids (BAs) in the serum was significantly altered in these individuals and marked by both elevated proportion and concentration of primary and conjugated BAs. Moreover, the NTCP deficiency led to increased levels of serum BAs, decreased levels of vitamin D, and aggravated the osteoporotic phenotype induced by estrogen withdrawal in mice. CONCLUSIONS Both mice and humans with NTCP deficiency presented hypercholanemia and were more prone to vitamin D deficiency and aggravated osteoporotic phenotype. Therefore, we recommend monitoring the levels of BAs and vitamin D, bone density, and abdominal ultrasounds in individuals with NTCP deficiency.
Collapse
Affiliation(s)
- Fangji Yang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenxiong Xu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lina Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Luo Yang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shu Zhu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenbin Wu
- Department of Spine Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuzhen Zhang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yutian Chong
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Liver Disease of Guangdong Province, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Liang Peng, ; Yutian Chong,
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Liver Disease of Guangdong Province, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Liang Peng, ; Yutian Chong,
| |
Collapse
|
10
|
Sun Z, Huang C, Shi Y, Wang R, Fan J, Yu Y, Zhang Z, Zhu K, Li M, Ni Q, Chen Z, Zheng M, Yang Z. Distinct Bile Acid Profiles in Patients With Chronic Hepatitis B Virus Infection Reveal Metabolic Interplay Between Host, Virus and Gut Microbiome. Front Med (Lausanne) 2021; 8:708495. [PMID: 34671614 PMCID: PMC8520922 DOI: 10.3389/fmed.2021.708495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022] Open
Abstract
Hepatitis B virus (HBV) can hijack the host bile acids (BAs) metabolic pathway during infection in cell and animal models. Additionally, microbiome was known to play critical role in the enterohepatic cycle of BAs. However, the impact of HBV infection and associated gut microbiota on the BA metabolism in chronic hepatitis B (CHB) patients is unknown. This study aimed to unveil the distinct BA profiles in chronic HBV infection (CHB) patients with no or mild hepatic injury, and to explore the relationship between HBV, microbiome and BA metabolism with clinical implications. Methods: Serum BA profiles were compared between CHB patients with normal ALT (CHB-NALT, n = 92), with abnormal ALT (CHB-AALT, n = 34) and healthy controls (HCs, n = 28) using UPLC-MS measurement. Hepatic gene expression in CHB patients were explored using previously published transcriptomic data. Fecal microbiome was compared between 30 CHB-NALT and 30 HCs using 16S rRNA sequencing, and key microbial function was predicted by PICRUSt analysis. Results: Significant higher percentage of conjugated BAs and primary BAs was found in CHB patients even without apparent liver injury. Combinatory BA features can discriminate CHB patients and HCs with high accuracy (AUC = 0.838). Up-regulation of BA importer Na+ taurocholate co-transporting peptide (NTCP) and down-regulation of bile salt export pump (BSEP) was found in CHB-NALT patients. The microbial diversity and abundance of Lactobacillus, Clostridium, Bifidobacterium were lower in CHB-NALT patients compared to healthy controls. Suppressed microbial bile salt hydrolases (BSH), 7-alpha-hydroxysteroid dehydrogenase (hdhA) and 3-dehydro-bile acid Delta 4, 6-reductase (BaiN) activity were found in CHB-NALT patients. Conclusion: This study provides new insight into the BA metabolism influenced both by HBV infection and associated gut microbiome modulations, and may lead to novel strategy for clinical management for chronic HBV infection.
Collapse
Affiliation(s)
- Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Chenjie Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.,Kidney Disease Center, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Yixian Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.,Department of Hepatology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Rusha Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jun Fan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ye Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhehua Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Kundan Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Minwei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Qin Ni
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhenggang Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Xu C, Bao Y, Zuo J, Li Y, Tang Y, Qu X, Ying H. Maternal chronic hepatitis B virus infection and the risk of preterm birth: A retrospective cohort analysis in Chinese women. J Viral Hepat 2021; 28:1422-1430. [PMID: 34342096 DOI: 10.1111/jvh.13585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/23/2021] [Indexed: 01/05/2023]
Abstract
The association between different clinical states of chronic HBV infection and preterm birth (PTB) is still controversial. A retrospective cohort study among 57,386 pregnant women was conducted to examine the impact of chronic HBsAg positive, both HBsAg and HBeAg positive, and chronic active hepatitis on pregnancy complications related to the overall PTB and its subtypes (spontaneous and iatrogenic). A total of 54,245 pregnancies were included in the final study cohort, among which 2,151(4.0%) pregnant women were HBsAg positive. The PTB rate was 6.0% (129/2151) for HBV-infected women while 4.5% (2319/52094) for those not. Compared with women not infected with HBV, multivariable-adjusted analyses showed HBV-infected women had a 33% higher risk of overall PTB (aRR 1.33 95%CI, 1.11-1.60), a 27% higher risk of spontaneous PTB (aRR 1.27, 95% CI, 1.02-1.57) and a 50% higher risk of iatrogenic PTB (aRR 1.50, 95%CI, 1.07-2.11). The PTB rate was 8.9% (35/395) for both HBsAg and HBeAg-positive women and 16.2% (22/136) for women with active chronic hepatitis. Multivariable-adjusted analyses showed women who were both HBsAg and HBeAg positive had a 47% higher risk of overall PTB (aRR 1.47, 95%CI, 1.04-2.09), a 2.03 times higher risk of spontaneous PTB (aRR 2.03, 95%CI, 1.38-2.99) and a 32% higher risk of iatrogenic PTB (aRR 1.32, 95%CI, 0.62-2.81), while women with chronic active hepatitis had a 3.84 times higher risk of overall PTB (aRR 3.84, 95%CI, 2.42-6.10), a 3.88 times higher risk of spontaneous PTB (aRR 3.88, 95%CI, 2.32-6.45) and a 3.01 times higher risk of iatrogenic PTB (aRR 3.01, 95%CI, 1.22-7.44). Different maternal clinical states of chronic HBV infection are independently associated with an increased risk of overall PTB and its subtypes (spontaneous and iatrogenic).
Collapse
Affiliation(s)
- Chuanlu Xu
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yirong Bao
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia Zuo
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuhong Li
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuping Tang
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxian Qu
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Ying
- Shanghai Key Laboratory of maternal fetal medicine, Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Zuccaro V, Asperges E, Colaneri M, Marvulli LN, Bruno R. HBV and HDV: New Treatments on the Horizon. J Clin Med 2021; 10:jcm10184054. [PMID: 34575165 PMCID: PMC8471459 DOI: 10.3390/jcm10184054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the accumulating knowledge, chronic hepatitis B (CHB) and HDV infection represent a global health problem, and there are still several critical issues, which frequently remain uncovered. In this paper, we provided an overview of the current therapeutic options and summarized the investigational therapies in the pipeline. Furthermore, we discussed some critical issues such as a “functional cure” approach, the futility of long-term NA therapy and the relevance of understanding drug actions and safety of antivirals, especially in special populations.
Collapse
Affiliation(s)
- Valentina Zuccaro
- U.O.C. Malattie Infettive I Fondazione IRCCS Policlinico San Matteo–Università di Pavia, 27100 Pavia, Italy; (E.A.); (M.C.); (L.N.M.); (R.B.)
- Correspondence: ; Tel.: +39-0382502660
| | - Erika Asperges
- U.O.C. Malattie Infettive I Fondazione IRCCS Policlinico San Matteo–Università di Pavia, 27100 Pavia, Italy; (E.A.); (M.C.); (L.N.M.); (R.B.)
| | - Marta Colaneri
- U.O.C. Malattie Infettive I Fondazione IRCCS Policlinico San Matteo–Università di Pavia, 27100 Pavia, Italy; (E.A.); (M.C.); (L.N.M.); (R.B.)
| | - Lea Nadia Marvulli
- U.O.C. Malattie Infettive I Fondazione IRCCS Policlinico San Matteo–Università di Pavia, 27100 Pavia, Italy; (E.A.); (M.C.); (L.N.M.); (R.B.)
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche–Università di Pavia, 27100 Pavia, Italy
| | - Raffaele Bruno
- U.O.C. Malattie Infettive I Fondazione IRCCS Policlinico San Matteo–Università di Pavia, 27100 Pavia, Italy; (E.A.); (M.C.); (L.N.M.); (R.B.)
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche–Università di Pavia, 27100 Pavia, Italy
| |
Collapse
|
13
|
Lyu J, Li H, Yin D, Zhao M, Sun Q, Guo M. Analysis of eight bile acids in urine of gastric cancer patients based on covalent organic framework enrichment coupled with liquid chromatography-tandem mass spectrometry. J Chromatogr A 2021; 1653:462422. [PMID: 34348207 DOI: 10.1016/j.chroma.2021.462422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Gastric carcinoma is one of the most common and deadly forms of cancer. Early detection is critical for successful treatment of gastric cancer, and examination of BAs in urine may provide a critical diagnostic tool for identifying gastric cancer at stages when it can still be cured. Bile acids (BAs) are a crucial toxic factor correlated with the injury of gastric mucosa and as such, quantifying the amount of BA in patient's urine could provide a new means to quickly and non-invasively identify the presence of gastric cancer in the early stages. Here, a covalent organic framework (COF) material synthesized on the basis of 1,3,5-tris(4-nitrophenyl)benzene (TAPB) and pyromellitic dianhydride (PMDA) was used as stationary phase for SPE column that was coupled to LC-MS/MS for quantitative analysis of eight BAs in human urine, including cholic acid (CA), deoxycholic acid (DCA), glycochenodeoxycholic acid (GCDCA), glycocholic acid (GCA), taurochenodeoxycholic acid (TCDCA), lithocholic acid (LCA), hyodeoxycholic acid (HDCA), and chenodeoxycholic acid (CDCA). The enrichment effect of synthesized COF material was better than commercial SPE and HLB column. The sensitivity can increase 9.37- to 54.30- fold (calculated by the ratio of peak area between before and after enrichment). The probable mechanism is due to the great porosity and the similar polarity with BAs of the COF material. By compared with previous literatures, our method had the minimum limit of detection, which achieved 46.40, 25.75, 47.40, 47.37, 30.42, and 33.92 pg /mL, respectively, for GCA, GCDCA, CA, CDCA, HDCA and DCA after enrichment. These eight BAs also accomplished excellent linearity from 0.34 to 10,000 ng/mL. This material was successfully applied in the measurements of these six BAs in human urine from 76 gastric cancer patients and 32 healthy people. Compared to healthy people, levels of CA, CDCA, DCA, and HDCA were significantly elevated and levels of GCDCA were depressed, respectively, in gastric cancer patients. Our work suggests that these acids may act as potential biomarkers for gastric cancer and our framework provides a method for "non-invasive" diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Jinxiu Lyu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Haijuan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Dengyang Yin
- Jingjiang People's Hospital, Taizhou, Jiangsu, 214500, China
| | - Meng Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Qiang Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Mengzhe Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
14
|
Abstract
Bulevirtide (Hepcludex®), a first-in-class entry inhibitor, is being developed by MYR GmbH for the treatment of chronic hepatitis delta virus (HDV) and chronic hepatitis B virus (HBV) infections. Bulevirtide was recently approved in the European Union (EU) for the treatment of chronic HDV infection in HDV RNA positive adult patients with compensated liver disease. This article summarizes the milestones in the development of bulevirtide leading to this first approval for chronic HDV.
Collapse
|
15
|
Jiang R, Wang T, Yao Y, Zhou F, Huang X. Hepatitis B infection and intrahepatic cholestasis of pregnancy: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e21416. [PMID: 32756142 PMCID: PMC7402766 DOI: 10.1097/md.0000000000021416] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Viral hepatitis type B is caused by hepatitis B virus (HBV) infection. Several studies have linked HBV infection to a higher risk of developing intrahepatic cholestasis of pregnancy (ICP), although some give contradictory results. To investigate the association and estimated risk of ICP in patients with HBV infection, we conducted this meta-analysis to summarize all available evidence. METHODS This study consists of 2 meta-analyses. A literature search was performed using MEDLINE and EMBASE from inception to July 2019. The first study included studies that reported associations between HBV infection and the risk of ICP. The second analysis included studies comparing the risk of HBV infection in ICP patients with those without ICP. Odds ratios (OR) and 95% confidence intervals (CI) were calculated using a random-effect, inverse variance method. RESULTS Four studies were included in both analyses. The OR of ICP in HBV-infected pregnant women compared with non-HBV pregnant women was 1.68 (95% CI 1.43-1.97; I = 0%). The OR of HBV infection among ICP patients compared with non-ICP patients was 1.70 (95% CI 1.44-2.01; I = 0%). CONCLUSIONS Our meta-analysis demonstrates not only a higher risk of ICP among HBV-infected pregnant women but also an increased risk of HBV infection among ICP patients. These findings suggest that HBV is a high-risk factor for ICP and screening for hepatitis B in women with ICP symptoms may be beneficial.
Collapse
|
16
|
Dosedělová V, Itterheimová P, Kubáň P. Analysis of bile acids in human biological samples by microcolumn separation techniques: A review. Electrophoresis 2020; 42:68-85. [PMID: 32645223 DOI: 10.1002/elps.202000139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022]
Abstract
Bile acids are a group of compounds essential for lipid digestion and absorption with a steroid skeleton and a carboxylate side chain usually conjugated to glycine or taurine. Bile acids are regulatory molecules for a number of metabolic processes and can be used as biomarkers of various disorders. Since the middle of the twentieth century, the detection of bile acids has evolved from simple qualitative analysis to accurate quantification in complicated mixtures. Advanced methods are required to characterize and quantify individual bile acids in these mixtures. This article overviews the literature from the last two decades (2000-2020) and focuses on bile acid analysis in various human biological samples. The methods for sample preparation, including the sample treatment of conventional (blood plasma, blood serum, and urine) and unconventional samples (bile, saliva, duodenal/gastric juice, feces, etc.) are shortly discussed. Eventually, the focus is on novel analytical approaches and methods for each particular biological sample, providing an overview of the microcolumn separation techniques, such as high-performance liquid chromatography, gas chromatography, and capillary electrophoresis, used in their analysis. This is followed by a discussion on selected clinical applications.
Collapse
Affiliation(s)
- Věra Dosedělová
- Department of Bioanalytical Instrumentation, CEITEC Masaryk University, Brno, Czech Republic
| | - Petra Itterheimová
- Department of Bioanalytical Instrumentation, CEITEC Masaryk University, Brno, Czech Republic
| | - Petr Kubáň
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| |
Collapse
|
17
|
Yee WLS, Drum CL. Increasing Complexity to Simplify Clinical Care: High Resolution Mass Spectrometry as an Enabler of AI Guided Clinical and Therapeutic Monitoring. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Wei Loong Sherman Yee
- Yong Loo Lin School of MedicineDepartment of MedicineNational University of Singapore Singapore 119077 Singapore
- Cardiovascular Research Institute (CVRI)National University Health System Singapore 119228 Singapore
| | - Chester Lee Drum
- Yong Loo Lin School of MedicineDepartment of MedicineNational University of Singapore Singapore 119077 Singapore
- Cardiovascular Research Institute (CVRI)National University Health System Singapore 119228 Singapore
- Yong Loo Lin School of MedicineDepartment of BiochemistryNational University of Singapore Singapore 119077 Singapore
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 119077 Singapore
| |
Collapse
|
18
|
Groeneweg S, van Geest FS, Peeters RP, Heuer H, Visser WE. Thyroid Hormone Transporters. Endocr Rev 2020; 41:5637505. [PMID: 31754699 DOI: 10.1210/endrev/bnz008] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
Abstract
Thyroid hormone transporters at the plasma membrane govern intracellular bioavailability of thyroid hormone. Monocarboxylate transporter (MCT) 8 and MCT10, organic anion transporting polypeptide (OATP) 1C1, and SLC17A4 are currently known as transporters displaying the highest specificity toward thyroid hormones. Structure-function studies using homology modeling and mutational screens have led to better understanding of the molecular basis of thyroid hormone transport. Mutations in MCT8 and in OATP1C1 have been associated with clinical disorders. Different animal models have provided insight into the functional role of thyroid hormone transporters, in particular MCT8. Different treatment strategies for MCT8 deficiency have been explored, of which thyroid hormone analogue therapy is currently applied in patients. Future studies may reveal the identity of as-yet-undiscovered thyroid hormone transporters. Complementary studies employing animal and human models will provide further insight into the role of transporters in health and disease. (Endocrine Reviews 41: 1 - 55, 2020).
Collapse
Affiliation(s)
- Stefan Groeneweg
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ferdy S van Geest
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Robin P Peeters
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - W Edward Visser
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands Academic Center for Thyroid Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Goh B, Choi J, Kang JA, Park SG, Seo J, Kim TY. Development of a mass spectrometric screening assay for hepatitis B virus entry inhibitors. J Pharm Biomed Anal 2019; 178:112959. [PMID: 31722821 DOI: 10.1016/j.jpba.2019.112959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 12/24/2022]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) involved in bile acid transport in the liver is an entry receptor of hepatitis B virus (HBV). In the present study, we introduce a mass spectrometric screening assay for targeting HBV entry inhibitors that can reduce NTCP transporter activity by employing taurocholic acid (TCA) labeled with stable isotope (2,2,4,4-d4-TCA, d4-TCA) and NTCP-overexpressing human liver cancer cell lines such as HepG2 and Huh-7. The accuracy and reliability of the proposed mass spectrometric NTCP activity assay have been validated with known HBV inhibitors including cyclosporine A (CsA) and pre-S1 peptide (PreS/2-48Myr or myrcludex B analog) that suppress the entry of HBV into hepatocytes by targeting NTCP. For the inhibitor screening assay, NTCP-overexpressing HepG2 or Huh-7 cells are treated with either a combination of TCA and an inhibitor (CsA or PreS/2-48Myr) or d4-TCA alone to serve as a reference. The activity of an HBV inhibitor is determined by relative quantification between TCA and d4-TCA in a 1:1 mixture of inhibitor-treated cells and untreated control cells using liquid chromatography-mass spectrometry. With our new approach, the half maximal inhibitory concentration (IC50) values for CsA and PreS/2-48Myr have been determined at micromolar and nanomolar concentrations, respectively, which is consistent with the previous results obtained with other conventional HBV entry inhibitor assay methods. Our assay method does not require HBV infection or radioactive 3H-TCA and provides a facile way to identify viral entry inhibitors via measuring bile acid transport activity of NTCP.
Collapse
Affiliation(s)
- Byoungsook Goh
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jieun Choi
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jung-Ah Kang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jiwon Seo
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| | - Tae-Young Kim
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea; School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| |
Collapse
|
20
|
Dutta M, Cai J, Gui W, Patterson AD. A review of analytical platforms for accurate bile acid measurement. Anal Bioanal Chem 2019; 411:4541-4549. [PMID: 31127337 DOI: 10.1007/s00216-019-01890-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/19/2019] [Accepted: 04/30/2019] [Indexed: 12/23/2022]
Abstract
Bile acids are acidic steroids which help in lipid absorption, act as signaling molecules, and are key intermediate molecules between host and gut microbial metabolism. Perturbations in the circulating bile acid pool can lead to dysregulated metabolic and immunological function which may be associated with liver and intestinal disease. Bile acids have chemically diverse structures and are present in a broad range of concentrations in a wide variety of samples with complex biological matrices. Advanced analytical methods are therefore required to identify and accurately quantify individual bile acids. Though enzymatic determination of total bile acid is most popular in clinical laboratories, these methods provide limited information about individual bile acids. Advanced analytical methods such as gas chromatography- and liquid chromatography-mass spectrometry and nuclear magnetic resonance spectroscopy are highly informative techniques which help in identification and quantification of individual bile acids in complex biological matrices. Here, we review the detection technologies currently used for bile acid identification and quantification. We further discuss the advantages and disadvantages of these analytical techniques with respect to sensitivity, specificity, robustness, and ease of use. Graphical abstract.
Collapse
Affiliation(s)
- Mainak Dutta
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani, Dubai Campus, Dubai International Academic City, Dubai, UAE
| | - Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 322 Life Sciences Building, University Park, State College, PA, 16802, USA
| | - Wei Gui
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 322 Life Sciences Building, University Park, State College, PA, 16802, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 322 Life Sciences Building, University Park, State College, PA, 16802, USA.
| |
Collapse
|
21
|
Müller F, Sharma A, König J, Fromm MF. Biomarkers for In Vivo Assessment of Transporter Function. Pharmacol Rev 2018; 70:246-277. [PMID: 29487084 DOI: 10.1124/pr.116.013326] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-drug interactions are a major concern not only during clinical practice, but also in drug development. Due to limitations of in vitro-in vivo predictions of transporter-mediated drug-drug interactions, multiple clinical Phase I drug-drug interaction studies may become necessary for a new molecular entity to assess potential drug interaction liabilities. This is a resource-intensive process and exposes study participants, who frequently are healthy volunteers without benefit from study treatment, to the potential risks of a new drug in development. Therefore, there is currently a major interest in new approaches for better prediction of transporter-mediated drug-drug interactions. In particular, researchers in the field attempt to identify endogenous compounds as biomarkers for transporter function, such as hexadecanedioate, tetradecanedioate, coproporphyrins I and III, or glycochenodeoxycholate sulfate for hepatic uptake via organic anion transporting polypeptide 1B or N1-methylnicotinamide for multidrug and toxin extrusion protein-mediated renal secretion. We summarize in this review the currently proposed biomarkers and potential limitations of the substances identified to date. Moreover, we suggest criteria based on current experiences, which may be used to assess the suitability of a biomarker for transporter function. Finally, further alternatives and supplemental approaches to classic drug-drug interaction studies are discussed.
Collapse
Affiliation(s)
- Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Ashish Sharma
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (F.M., J.K., M.F.F.); and Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riß, Germany (F.M., A.S.)
| |
Collapse
|
22
|
Zhao K, Liu S, Chen Y, Yao Y, Zhou M, Yuan Y, Wang Y, Pei R, Chen J, Hu X, Zhou Y, Zhao H, Lu M, Wu C, Chen X. Upregulation of HBV transcription by sodium taurocholate cotransporting polypeptide at the postentry step is inhibited by the entry inhibitor Myrcludex B. Emerg Microbes Infect 2018; 7:186. [PMID: 30459339 PMCID: PMC6246608 DOI: 10.1038/s41426-018-0189-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/30/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for hepatitis B virus (HBV) entry. However, little is known regarding whether NTCP is involved in regulating the postentry steps of the HBV life cycle. Here, we found that NTCP expression upregulated HBV transcription at the postentry step and that the NTCP-targeting entry inhibitor Myrcludex B (MyrB) effectively suppressed HBV transcription both in an HBV in vitro infection system and in mice hydrodynamically injected with an HBV expression plasmid. Mechanistically, NTCP upregulated HBV transcription via farnesoid X receptor α (FxRα)-mediated activation of the HBV EN2/core promoter at the postentry step in a manner that was dependent on the bile acid (BA)-transport function of NTCP, which was blocked by MyrB. Our findings uncover a novel role for NTCP in the HBV life cycle and provide a reference for the use of novel NTCP-targeting entry inhibitors to suppress HBV infection and replication.
Collapse
Affiliation(s)
- Kaitao Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shuhui Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yingshan Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yongxuan Yao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ming Zhou
- Shenzhen Xenotransplantation Research and Development Center, State and Local Joint Cancer Genome Clinical Application of Key Technology Laboratory, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 518035, Shenzhen, China
| | - Yifei Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yun Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Jizheng Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - He Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, 45147, Essen, Germany
| | - Chunchen Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.
| | - Xinwen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
23
|
Fujiwara R, Haag M, Schaeffeler E, Nies AT, Zanger UM, Schwab M. Systemic regulation of bilirubin homeostasis: Potential benefits of hyperbilirubinemia. Hepatology 2018; 67:1609-1619. [PMID: 29059457 DOI: 10.1002/hep.29599] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/11/2017] [Accepted: 10/13/2017] [Indexed: 12/17/2022]
Abstract
Neurotoxic bilirubin is the end product of heme catabolism in mammals. Bilirubin is solely conjugated by uridine diphospho-glucuronosyltransferase 1A1, which is a membrane-bound enzyme that catalyzes the transfer of glucuronic acid. Due to low function of hepatic and intestinal uridine diphospho-glucuronosyltransferase 1A1 during the neonatal period, human neonates develop mild to severe physiological hyperbilirubinemia. Accumulation of bilirubin in the brain leads to the onset of irreversible brain damage, termed kernicterus. Breastfeeding is one of the most significant factors that increase the risk of developing kernicterus in infants. Why does this most natural way of feeding increase the risk of brain damage or even death? This question leads to the hypothesis that breast milk-induced hyperbilirubinemia might bring certain benefits that outweigh those risks. While bilirubin is neurotoxic and cytotoxic, this compound is also a potent antioxidant. There are studies showing improved clinical conditions in patients with hyperbilirubinemia. Accumulating evidence also shows that genetic polymorphisms linked to hyperbilirubinemia are beneficial against various diseases. In this review article, we first introduce the production, metabolism, and transport of bilirubin. We then discuss the potential benefits of neonatal and adult hyperbilirubinemia. Finally, epigenetic factors as well as metabolomic information associated with hyperbilirubinemia are described. This review article advances the understanding of the physiological importance of the paradoxical compound bilirubin. (Hepatology 2018;67:1609-1619).
Collapse
Affiliation(s)
- Ryoichi Fujiwara
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany.,Department of Pharmaceutics, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany.,Department of Clinical Pharmacology, University Hospital, Tuebingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Slijepcevic D, Roscam Abbing RL, Katafuchi T, Blank A, Donkers JM, van Hoppe S, de Waart DR, Tolenaars D, van der Meer JH, Wildenberg M, Beuers U, Oude Elferink RP, Schinkel AH, van de Graaf SF. Hepatic uptake of conjugated bile acids is mediated by both sodium taurocholate cotransporting polypeptide and organic anion transporting polypeptides and modulated by intestinal sensing of plasma bile acid levels in mice. Hepatology 2017; 66:1631-1643. [PMID: 28498614 PMCID: PMC5698707 DOI: 10.1002/hep.29251] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 04/07/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022]
Abstract
UNLABELLED The Na+ -taurocholate cotransporting polypeptide (NTCP/SLC10A1) is believed to be pivotal for hepatic uptake of conjugated bile acids. However, plasma bile acid levels are normal in a subset of NTCP knockout mice and in mice treated with myrcludex B, a specific NTCP inhibitor. Here, we elucidated which transport proteins mediate the hepatic uptake of conjugated bile acids and demonstrated intestinal sensing of elevated bile acid levels in plasma in mice. Mice or healthy volunteers were treated with myrcludex B. Hepatic bile acid uptake kinetics were determined in wild-type (WT), organic anion transporting polypeptide (OATP) knockout mice (lacking Slco1a/1b isoforms), and human OATP1B1-transgenic mice. Effects of fibroblast growth factor 19 (FGF19) on hepatic transporter mRNA levels were assessed in rat hepatoma cells and in mice by peptide injection or adeno-associated virus-mediated overexpression. NTCP inhibition using myrcludex B had only moderate effects on bile acid kinetics in WT mice, but completely inhibited active transport of conjugated bile acid species in OATP knockout mice. Cholesterol 7α-hydroxylase Cyp7a1 expression was strongly down-regulated upon prolonged inhibition of hepatic uptake of conjugated bile acids. Fgf15 (mouse counterpart of FGF19) expression was induced in hypercholanemic OATP and NTCP knockout mice, as well as in myrcludex B-treated cholestatic mice, whereas plasma FGF19 was not induced in humans treated with myrcludex B. Fgf15/FGF19 expression was induced in polarized human enterocyte-models and mouse organoids by basolateral incubation with a high concentration (1 mM) of conjugated bile acids. CONCLUSION NTCP and OATPs contribute to hepatic uptake of conjugated bile acids in mice, whereas the predominant uptake in humans is NTCP mediated. Enterocytes sense highly elevated levels of (conjugated) bile acids in the systemic circulation to induce FGF15/19, which modulates hepatic bile acid synthesis and uptake. (Hepatology 2017;66:1631-1643).
Collapse
Affiliation(s)
- Davor Slijepcevic
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands
| | | | | | - Antje Blank
- Department of Clinical Pharmacology and PharmacoepidemiologyHeidelberg University HospitalHeidelbergGermany,German Center for Infection Research (DZIF)Heidelberg Partner SiteHeidelbergGermany
| | - Joanne M. Donkers
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands
| | - Stéphanie van Hoppe
- Division of Molecular Oncologythe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Dirk. R. de Waart
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands
| | - Dagmar Tolenaars
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands
| | | | - Manon Wildenberg
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands,Department of Gastroenterology and HepatologyAcademic Medical CenterAmsterdamThe Netherlands
| | - Ulrich Beuers
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands,Department of Gastroenterology and HepatologyAcademic Medical CenterAmsterdamThe Netherlands
| | - Ronald P.J. Oude Elferink
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands,Department of Gastroenterology and HepatologyAcademic Medical CenterAmsterdamThe Netherlands
| | - Alfred H. Schinkel
- Division of Molecular Oncologythe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Stan F.J. van de Graaf
- Tytgat Institute for Liver and Intestinal ResearchAcademic Medical CenterAmsterdamThe Netherlands,Department of Gastroenterology and HepatologyAcademic Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
25
|
Peng Z, Zhang Q, Mao Z, Wang J, Liu C, Lin X, Li X, Ji W, Fan J, Wang M, Su C. A rapid quantitative analysis of bile acids, lysophosphatidylcholines and polyunsaturated fatty acids in biofluids based on ultraperformance liquid chromatography coupled with triple quadrupole tandem massspectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1068-1069:343-351. [DOI: 10.1016/j.jchromb.2017.10.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/24/2022]
|
26
|
Lempp FA, Urban S. Hepatitis Delta Virus: Replication Strategy and Upcoming Therapeutic Options for a Neglected Human Pathogen. Viruses 2017; 9:E172. [PMID: 28677645 PMCID: PMC5537664 DOI: 10.3390/v9070172] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022] Open
Abstract
The human Hepatitis Delta Virus (HDV) is unique among all viral pathogens. Encoding only one protein (Hepatitis Delta Antigen; HDAg) within its viroid-like self-complementary RNA, HDV constitutes the smallest known virus in the animal kingdom. To disseminate in its host, HDV depends on a helper virus, the human Hepatitis B virus (HBV), which provides the envelope proteins required for HDV assembly. HDV affects an estimated 15-20 million out of the 240 million chronic HBV-carriers and disperses unequally in disparate geographical regions of the world. The disease it causes (chronic Hepatitis D) presents as the most severe form of viral hepatitis, leading to accelerated progression of liver dysfunction including cirrhosis and hepatocellular carcinoma and a high mortality rate. The lack of approved drugs interfering with specific steps of HDV replication poses a high burden for gaining insights into the molecular biology of the virus and, consequently, the development of specific novel medications that resiliently control HDV replication or, in the best case, functionally cure HDV infection or HBV/HDV co-infection. This review summarizes our current knowledge of HBV molecular biology, presents an update on novel cell culture and animal models to study the virus and provides updates on the clinical development of the three developmental drugs Lonafarnib, REP2139-Ca and Myrcludex B.
Collapse
Affiliation(s)
- Florian A Lempp
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany.
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
27
|
Blank A, Eidam A, Haag M, Hohmann N, Burhenne J, Schwab M, van de Graaf SFJ, Meyer MR, Maurer HH, Meier K, Weiss J, Bruckner T, Alexandrov A, Urban S, Mikus G, Haefeli WE. The NTCP-inhibitor Myrcludex B: Effects on Bile Acid Disposition and Tenofovir Pharmacokinetics. Clin Pharmacol Ther 2017; 103:341-348. [DOI: 10.1002/cpt.744] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/10/2017] [Accepted: 05/13/2017] [Indexed: 12/26/2022]
Affiliation(s)
- A Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - A Eidam
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - M Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology; University of Tübingen; Stuttgart Germany
- German Center for Infection Research (DZIF); Tübingen Partner Site; Tübingen Germany
| | - N Hohmann
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - J Burhenne
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - M Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology; University of Tübingen; Stuttgart Germany
- German Center for Infection Research (DZIF); Tübingen Partner Site; Tübingen Germany
- Department of Clinical Pharmacology; University Hospital Tübingen; Tübingen Germany
- Department of Pharmacy and Biochemistry; University of Tübingen; Tübingen Germany
| | - SFJ van de Graaf
- Tytgat Institute for Liver and Intestinal Research & Department of Gastroenterology & Hepatology; Academic Medical Center; Amsterdam The Netherlands
| | - MR Meyer
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- Experimental and Clinical Toxicology; Saarland University; Homburg Germany
| | - HH Maurer
- Experimental and Clinical Toxicology; Saarland University; Homburg Germany
| | - K Meier
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - J Weiss
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - T Bruckner
- Institute of Medical Biostatistics and Medical Informatics; Heidelberg University Hospital; Heidelberg Germany
| | | | - S Urban
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
- Department of Infectious Diseases, Molecular Virology; Heidelberg University Hospital; Heidelberg Germany
| | - G Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| | - WE Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology; Heidelberg University Hospital; Heidelberg Germany
- German Center for Infection Research (DZIF); Heidelberg Partner Site; Heidelberg Germany
| |
Collapse
|
28
|
Leuthold P, Schaeffeler E, Winter S, Büttner F, Hofmann U, Mürdter TE, Rausch S, Sonntag D, Wahrheit J, Fend F, Hennenlotter J, Bedke J, Schwab M, Haag M. Comprehensive Metabolomic and Lipidomic Profiling of Human Kidney Tissue: A Platform Comparison. J Proteome Res 2017; 16:933-944. [DOI: 10.1021/acs.jproteome.6b00875] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Patrick Leuthold
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Florian Büttner
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Thomas E. Mürdter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Steffen Rausch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
- Department
of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | | | | | - Falko Fend
- Institute
of Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Jörg Hennenlotter
- Department
of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Jens Bedke
- Department
of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
- Department
of Clinical Pharmacology, University Hospital Tübingen, 72076 Tübingen, Germany
- Department
of Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
29
|
Wegner K, Just S, Gau L, Mueller H, Gérard P, Lepage P, Clavel T, Rohn S. Rapid analysis of bile acids in different biological matrices using LC-ESI-MS/MS for the investigation of bile acid transformation by mammalian gut bacteria. Anal Bioanal Chem 2016; 409:1231-1245. [PMID: 27822648 DOI: 10.1007/s00216-016-0048-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/22/2016] [Accepted: 10/21/2016] [Indexed: 01/15/2023]
Abstract
Bile acids are important signaling molecules that regulate cholesterol, glucose, and energy homoeostasis and have thus been implicated in the development of metabolic disorders. Their bioavailability is strongly modulated by the gut microbiota, which contributes to generation of complex individual-specific bile acid profiles. Hence, it is important to have accurate methods at hand for precise measurement of these important metabolites. Here, a rapid and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for simultaneous identification and quantitation of primary and secondary bile acids as well as their taurine and glycine conjugates was developed and validated. Applicability of the method was demonstrated for mammalian tissues, biofluids, and cell culture media. The analytical approach mainly consists of a simple and rapid liquid-liquid extraction procedure in presence of deuterium-labeled internal standards. Baseline separation of all isobaric bile acid species was achieved and a linear correlation over a broad concentration range was observed. The method showed acceptable accuracy and precision on intra-day (1.42-11.07 %) and inter-day (2.11-12.71 %) analyses and achieved good recovery rates for representative analytes (83.7-107.1 %). As a proof of concept, the analytical method was applied to mouse tissues and biofluids, but especially to samples from in vitro fermentations with gut bacteria of the family Coriobacteriaceae. The developed method revealed that the species Eggerthella lenta and Collinsella aerofaciens possess bile salt hydrolase activity, and for the first time that the species Enterorhabdus mucosicola is able to deconjugate and dehydrogenate primary bile acids in vitro.
Collapse
Affiliation(s)
- Katrin Wegner
- University of Hamburg, Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany
| | - Sarah Just
- Junior Research Group Intestinal Microbiome, ZIEL-Institute for Food Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Laura Gau
- University of Hamburg, Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany
| | - Henrike Mueller
- University of Hamburg, Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Thomas Clavel
- Junior Research Group Intestinal Microbiome, ZIEL-Institute for Food Health, Technical University of Munich, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Sascha Rohn
- University of Hamburg, Hamburg School of Food Science, Institute of Food Chemistry, Grindelallee 117, 20146, Hamburg, Germany.
| |
Collapse
|
30
|
Lempp FA, Ni Y, Urban S. Hepatitis delta virus: insights into a peculiar pathogen and novel treatment options. Nat Rev Gastroenterol Hepatol 2016; 13:580-9. [PMID: 27534692 DOI: 10.1038/nrgastro.2016.126] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chronic hepatitis D is the most severe form of viral hepatitis, affecting ∼20 million HBV-infected people worldwide. The causative agent, hepatitis delta virus (HDV), is a unique human pathogen: it is the smallest known virus; it depends on HBV to disseminate its viroid-like RNA; it encodes only one protein (HDAg), which has both structural and regulatory functions; and it replicates using predominantly host proteins. The failure of HBV-specific nucleoside analogues to suppress the HBV helper function, and the limitations of experimental systems to study the HDV life cycle, have impeded the development of HDV-specific drugs. Thus, the only clinical regimen for HDV is IFNα, which shows some efficacy but long-term virological responses are rare. Insights into the receptor-mediated entry of HDV, and the observation that HDV assembly requires farnesyltransferase, have enabled novel therapeutic strategies to be developed. Interference with entry, for example through blockade of the HBV-HDV-specific receptor sodium/taurocholate cotransporting polypeptide NTCP by Myrcludex B, and inhibition of assembly by blockade of farnesyltransferase using lonafarnib or nucleic acid polymers such as REP 2139-Ca, have shown promising results in phase II studies. In this Review, we summarize our knowledge of HDV epidemiology, pathogenesis and molecular biology, with a particular emphasis on possible future developments.
Collapse
Affiliation(s)
- Florian A Lempp
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
31
|
Bogomolov P, Alexandrov A, Voronkova N, Macievich M, Kokina K, Petrachenkova M, Lehr T, Lempp FA, Wedemeyer H, Haag M, Schwab M, Haefeli WE, Blank A, Urban S. Treatment of chronic hepatitis D with the entry inhibitor myrcludex B: First results of a phase Ib/IIa study. J Hepatol 2016; 65:490-8. [PMID: 27132170 DOI: 10.1016/j.jhep.2016.04.016] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/19/2016] [Accepted: 04/19/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS The therapeutic option for patients with chronic hepatitis delta virus infection (CHD) is limited to interferon alpha with rare curative outcome. Myrcludex B is a first-in-class entry inhibitor inactivating the hepatitis B virus (HBV) and hepatitis D virus (HDV) receptor sodium taurocholate co-transporting polypeptide. We report the interim results of a pilot trial on chronically infected HDV patients treated with myrcludex B, or pegylated interferon alpha (PegIFNα-2a) or their combination. METHODS Twenty-four patients with CHD infection were equally randomized (1:1:1) to receive myrcludex B, or PegIFNα-2a or their combination. Patients were evaluated for virological and biochemical response and tolerability of the study drugs at weeks 12 and 24. RESULTS Myrcludex B was well tolerated and no serious adverse event occurred. Although hepatitis B surface antigen levels remained unchanged, HDV RNA significantly declined at week 24 in all cohorts. HDV RNA became negative in two patients each in the Myrcludex B and PegIFNα-2a cohorts, and in five patients of the Myrcludex B+PegIFNα-2a cohort. ALT decreased significantly in the Myrcludex B cohort (six of eight patients), and HBV DNA was significantly reduced at week 24 in the Myrcludex B+PegIFNα-2a cohort. Virus kinetic modeling suggested a strong synergistic effect of myrcludex B and PegIFNα-2a on both HDV and HBV. CONCLUSIONS Myrcludex B showed a strong effect on HDV RNA serum levels and induced ALT normalization under monotherapy. Synergistic antiviral effects on HDV RNA and HBV DNA in the Myr-IFN cohort indicated a benefit of the combination of entry inhibition with PegIFNα-2a to treat CHD patients. LAY SUMMARY Myrcludex B is a new drug to treat hepatitis B and D infection. After 24weeks of treatment with myrcludex B and/or pegylated interferon α-2a, HDV R NA, a relevant marker for hepatitis D infection, decreased in all patients with chronic hepatitis B and D. Two of eight patients which received either myrcludex B or pegylated interferon α-2a, became negative for HDV RNA, and five of seven patients who received both drugs at the same time became negative. The drug was well tolerated.
Collapse
Affiliation(s)
- Pavel Bogomolov
- Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2 Schepkina str., 129110 Moscow, Russia; Centrosoyuz Clinical Hospital, 57 Gilyarovskogo str., Moscow 129110, Russia
| | | | - Natalia Voronkova
- Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2 Schepkina str., 129110 Moscow, Russia; Centrosoyuz Clinical Hospital, 57 Gilyarovskogo str., Moscow 129110, Russia
| | - Maria Macievich
- Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2 Schepkina str., 129110 Moscow, Russia; Centrosoyuz Clinical Hospital, 57 Gilyarovskogo str., Moscow 129110, Russia
| | - Ksenia Kokina
- Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2 Schepkina str., 129110 Moscow, Russia; Centrosoyuz Clinical Hospital, 57 Gilyarovskogo str., Moscow 129110, Russia
| | - Maria Petrachenkova
- Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2 Schepkina str., 129110 Moscow, Russia; Centrosoyuz Clinical Hospital, 57 Gilyarovskogo str., Moscow 129110, Russia
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123 Saarbrücken, Germany
| | - Florian A Lempp
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstraße 112, 70376 Stuttgart, Germany; University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Tübingen Partner Site, E.-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstraße 112, 70376 Stuttgart, Germany; University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Tübingen Partner Site, E.-Aulhorn-Str. 6, 72076 Tübingen, Germany; Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany; Department of Pharmacy and Biochemistry, University of Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Walter E Haefeli
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Antje Blank
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | - Stephan Urban
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Blank A, Markert C, Hohmann N, Carls A, Mikus G, Lehr T, Alexandrov A, Haag M, Schwab M, Urban S, Haefeli WE. First-in-human application of the novel hepatitis B and hepatitis D virus entry inhibitor myrcludex B. J Hepatol 2016; 65:483-9. [PMID: 27132172 DOI: 10.1016/j.jhep.2016.04.013] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/27/2015] [Accepted: 04/19/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Myrcludex B is a first-in-class compound, which blocks entry of hepatitis B and D virus into hepatocytes in vitro and in animal models. Based on the required preclinical data we aimed to translate this compound into the first application in humans. METHODS Single ascending doses of myrcludex B, a 47 amino acid peptide, were administered up to 20mg intravenously and 10mg subcutaneously in a prospective open first-in-human, phase I clinical trial to 36 healthy volunteers. Safety, tolerability and plasma concentrations of myrcludex B were assessed and a pharmacokinetic model was derived. RESULTS Myrcludex B was well tolerated and no serious or relevant AEs representing off-target effects, and no immunogenic effects were observed up to the highest applied dose of 20mg (intravenously). Myrcludex B showed dose-dependent pharmacokinetics, best described by a 2-compartment target-mediated drug disposition model. Bioavailability of the subcutaneous application was large (85%). Interindividual variability was moderate. The pharmacokinetic model suggested that subcutaneous doses of 10mg and above reach a target saturation of over 80% for at least 15h. CONCLUSIONS Myrcludex B showed excellent tolerability up to high doses. Pharmacologic properties followed a 2-compartment target-mediated drug disposition model. These findings are vital for planning of further multiple dose efficacy trials in patients. LAY SUMMARY After showing antiviral activity in cell culture and animal models, myrcludex B, a new drug intended for the treatment of hepatitis B and D, has been administered the first time in humans. Healthy volunteers received the drug intravenously and subcutaneously up to high doses (20mg). The drug was well tolerated and the characteristics of the drug determining its way in the human body could be described. These results will allow testing myrcludex B in hepatitis B and D patients.
Collapse
Affiliation(s)
- Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | - Christoph Markert
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Nicolas Hohmann
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Alexandra Carls
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123 Saarbrücken, Germany
| | | | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstraße 112, 70376 Stuttgart, Germany; University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Tübingen Partner Site, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstraße 112, 70376 Stuttgart, Germany; University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Tübingen Partner Site, Tübingen, Germany; Department of Clinical Pharmacology, University Hospital Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
| | - Stephan Urban
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany; Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, 69120 Heidelberg, Germany
| | - Walter E Haefeli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg Partner Site, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
33
|
Pham HT, Arnhard K, Asad YJ, Deng L, Felder TK, St. John-Williams L, Kaever V, Leadley M, Mitro N, Muccio S, Prehn C, Rauh M, Rolle-Kampczyk U, Thompson JW, Uhl O, Ulaszewska M, Vogeser M, Wishart DS, Koal T. Inter-Laboratory Robustness of Next-Generation Bile Acid Study in Mice and Humans: International Ring Trial Involving 12 Laboratories. ACTA ACUST UNITED AC 2016; 1:129-142. [DOI: 10.1373/jalm.2016.020537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/01/2016] [Indexed: 11/06/2022]
|
34
|
Gao J, Xu B, Zhang X, Cui Y, Deng L, Shi Z, Shao Y, Ding M. Association between serum bile acid profiles and gestational diabetes mellitus: A targeted metabolomics study. Clin Chim Acta 2016; 459:63-72. [PMID: 27246871 DOI: 10.1016/j.cca.2016.05.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/10/2016] [Accepted: 05/27/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Given the potential influence of aberrant bile acid metabolism on glucose homeostasis, we hypothesized that serum bile acid metabolism is altered in gestational diabetes mellitus (GDM). We characterized the metabolic profiling changes of serum bile acids in GDM and to find the potential biomarkers for the diagnosis and differential diagnosis of GDM. METHODS Based on ultrahigh performance liquid chromatography/hybrid quadrupole time-of-flight mass spectrometry, a targeted metabolomics study that involved targeted and untargeted screening techniques was performed to explore the changes in serum bile acid metabolism of GDM cases, intrahepatic cholestasis of pregnancy (ICP) cases and healthy controls. RESULTS There were 3 significantly different profiling of serum bile acids for GDM, ICP and controls. Compared to the controls, GDM individuals demonstrated significant increases in 8 bile acid species, including 2 dihydroxy conjugated, 1 trihydroxy unconjugated and 5 sulfated bile acids. β-muricholic acid (β-MCA) and di-2 were well-suited to use as the metabolic markers for the diagnosis and differential diagnosis of GDM, respectively. CONCLUSIONS These preliminary findings revealed the protective effect of body against cytotoxicity via elimination of increased sulfated bile acids and aberrant enzyme activity participated in the cycle β-MCA→hyodeoxycholic acid (HDCA) of the bile acid metabolism pathway for the women with GDM, which gave us further insights into the etiology and pathophysiology of GDM.
Collapse
Affiliation(s)
- Jieying Gao
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Biao Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaoqing Zhang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yue Cui
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Linlin Deng
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhenghu Shi
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Shao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Min Ding
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|